Use of a physiologically-based pharmacokinetic model to explore the potential disparity in nicotine disposition between adult and adolescent nonhuman primates

https://doi.org/10.1016/j.taap.2019.114826Get rights and content

Highlights

  • A PBPK model was developed for nicotine in adult and juvenile monkeys

  • Juvenile monkeys clear nicotine and cotinine faster than adults

  • Age-dependent differences in monkeys may help inform nicotine kinetics in humans

Abstract

The widespread use and high abuse liability of tobacco products has received considerable public health attention, in particular for youth, who are vulnerable to nicotine addiction. In this study, adult and adolescent squirrel monkeys were used to evaluate age-related metabolism and pharmacokinetics of nicotine after intravenous administration. A physiologically-based pharmacokinetic (PBPK) model was created to characterize the pharmacokinetic behaviors of nicotine and its metabolites, cotinine, trans-3′-hydroxycotinine (3′-OH cotinine), and trans-3′-hydroxycotinine glucuronide (3′-OH cotinine glucuronide) for both adult and adolescent squirrel monkeys. The PBPK nicotine model was first calibrated for adult squirrel monkeys utilizing in vitro nicotine metabolic data, plasma concentration-time profiles and cumulative urinary excretion data for nicotine and metabolites. Further model refinement was conducted when the calibrated adult model was scaled to the adolescents, because adolescents appeared to clear nicotine and cotinine more rapidly relative to adults. More specifically, the resultant model parameters representing systemic clearance of nicotine and cotinine for adolescent monkeys were approximately two- to three-fold of the adult values on a per body weight basis. The nonhuman primate PBPK model in general captured experimental observations that were used for both model calibration and evaluation, with acceptable performance metrics for precision and bias. The model also identified differences in nicotine pharmacokinetics between adolescent and adult nonhuman primates which might also be present in humans.

Introduction

Nicotine (C10H14N2, CAS number 54-11-5, Molecular weight 162.23), though not the direct cause for most tobacco related diseases, is responsible for the addictive effects of tobacco products. Tobacco use remains the leading cause of preventable disease and deaths per year in the United States (FDA, 2017). Of particular concern are youth, in light of the potential increased risk for addiction and health issues. Both animal studies using adolescent (postnatal day, PND25) and adult (PND85) male Sprague-Dawley rats (Gellner et al., 2016), early adolescent (PND28), late adolescent (PND38), and adult (PND90) male Sprague-Dawley rats (Belluzzi et al., 2004), as well as clinical data, indicate that adolescents display greater sensitivity to the reinforcing properties of nicotine and are more likely to become nicotine dependent compared to adults (Chen and Millar, 1998). Tobacco use is primarily initiated and established during adolescence.

Nicotine pharmacokinetics plays a critical role in nicotine addiction and nicotine replacement therapy (Le Houezec, 2003). Pharmacokinetic profiles of nicotine have been widely studied in adult humans. Given that nicotine is a weak base with a pKa of 8.0, absorption of nicotine across biological membranes relies on the pH of the environments, since nicotine does not rapidly cross membranes in its ionized state. Following absorption, nicotine is largely distributed into body tissues. The plasma protein binding of nicotine is <5%. Nicotine is extensively metabolized by the liver, with the majority (70–80%) converted to cotinine (C10H12N2O, CAS number 486-56-6, Molecular weight 176.22) via oxidation. In addition, nicotine is also excreted by the kidney via glomerular filtration and tubular secretion, accounting for 5% of the total clearance (Benowitz et al., 2009). The terminal elimination half-life of plasma nicotine is about 2 h in adult humans (Matta et al., 2007).

Urinary clearance rates (half-lives) of cotinine in newborns whose mothers smoked tobacco have been reported (Etzel et al., 1985; Dempsey et al., 2000). In other studies, infants and youth were exposed to second hand smoke, then placed in a smoke-free environment before urine collection, and the urinary clearance of cotinine was calculated from the collected samples (Collier et al., 1994; Leong et al., 1998). These reported urinary half-lives were quite variable and conflicting, reflecting the shortcomings in using these opportunistic methods. When deuterated cotinine was administered to youth aged 2.5 to 82.4 months, urinary half-life estimates for cotinine were measured without interference of low-level contamination. The results showed that mean half-lives of cotinine ranged from a low value of 13.7 h in infants under one year of age and 19.6 h for youth 5–6 years of age (creatinine uncorrected) (Dempsey et al., 2013). However, to the best of our knowledge, no plasma pharmacokinetic studies of nicotine in youth are available in the literature and the difference in the pharmacokinetic characteristics of nicotine and its metabolites between youth and adults remains unclear.

Among different experimental animal models, nicotine metabolism in nonhuman primates is more comparable to humans (Matta et al., 2007). Like humans, nicotine is mostly converted to cotinine via oxidation, referred to as C-oxidation, in nonhuman primates, as identified in African green monkeys (Vervets, Cercopithecus aethiops), with CYP2A6 (referred to as CYP2A6agm) as the primary enzyme and a minor contribution from CYP2B6 (referred to as CYP2B6agm) (Schoedel et al., 2003). CYP2A6 and CYP2B6 expression, substrate specificity, and mediation are similar between nonhuman primates and their human orthologues (Schoedel et al., 2003), making nonhuman primates a more appropriate model than other animal models for the investigation of human nicotine pharmacokinetics. In addition, similar to humans, approximately 80% of nicotine is metabolized to cotinine in nonhuman primates (Poole and Urwin, 1976). Comparable terminal elimination half-lives for nicotine and cotinine have been observed between nonhuman primates and humans (Hukkanen et al., 2005). The terminal elimination half-life for nicotine is 1.6 h for macaque monkeys comparable to 2 h for adult humans (Hukkanen et al., 2005). For cotinine, a primary metabolite that is either cleared in urine or metabolized in the liver, the terminal elimination half-life is 9.2 h in the macaque monkey and 13.3 h in adult humans (Seaton et al., 1991).

In the current study, a nonhuman primate squirrel monkey model was used to evaluate the plasma and urinary pharmacokinetics of nicotine in adolescents and adults after intravenous administration of nicotine. The adolescent squirrel monkeys (1 to 3 years old) used in this study reflect a human youth's age of 6 to 16 years (Rowe, 1996; Beer et al., 2017), an important phase for neural development and tobacco use initiation. Further, a physiologically-based pharmacokinetic (PBPK) model was developed to characterize the plasma pharmacokinetics and urinary excretion of nicotine and its metabolites, cotinine, trans-3′-hydroxycotinine (3′-OH cotinine, C10H12N2O2, CAS number 34834-67-8, Molecular weight 192.21), and trans-3′-hydroxycotinine glucuronide (3′-OH cotinine glucuronide, C16H20N2O8, CAS number 132929-88-5, Molecular weight 368.34) for both adult and adolescent squirrel monkeys. Nicotine, as a highly addictive chemical compound, is also well known to cause adverse effects on the heart, lung, reproductive system, and kidney (Mishra et al., 2015). Nicotine's major metabolite, cotinine, which has been historically considered less harmful than nicotine, has also been shown to adversely impact the heart and reproductive system (Bastianini et al., 2018). As such, both nicotine and cotinine are included in the current model. In addition, to better track mass balance, the two major inactive metabolites, 3′-OH cotinine and 3′-OH cotinine glucuronides are also described in the model.

While several PBPK models have been developed to describe the pharmacokinetics of nicotine and cotinine in humans and experimental animal models, the existing models focused primarily on adults (Plowchalk et al., 1992, Robinson et al., 1992, Teeguarden et al., 2013, Gajewska et al., 2014, Saylor and Zhang, 2016). Additionally, to date there have been no nicotine PBPK models created for adult and adolescent nonhuman primates. Such knowledge would provide critical information to help identify age-dependent differences in nicotine pharmacokinetics in humans.

Section snippets

Ethics statement

All animal procedures were approved by the US FDA's National Center for Toxicological Research Institutional Animal Care and Use Committee, which were performed in accordance with the Public Health Service Policy on Human Care and Use of Laboratory Animals.

Subjects

In total, 14 adult (between 3 and 5 years of age, 891.7-1142.2 g) and 8 adolescent (between 1 and 3 years of age, 630.3–815 g) male squirrel monkeys (Saimiri sciureus; University of Texas, MD Anderson Cancer Center) served as subjects. The

Nicotine

The hepatic C-oxidation of nicotine for adult squirrel monkeys was described with a Michaelis affinity constant Km_nic of 3909.9 ng/mL and an optimized Vmax_nicC of 10,000 μg/h/kg0.75 (Table 3). The conversion of nicotine to other metabolites in the liver was described using a composite clearance term, Knic_otherC, with a value of 0.85 L/h/kg0.75. The model predicted that 74% of nicotine is converted to cotinine in squirrel monkeys. Subsequently, an optimized clearance term, Kurine_nicC, with

Model development and uncertainties

With the availability of new data for nicotine and its metabolites in plasma and urine collected in adult and adolescent squirrel monkeys, a PBPK model was created for the first time to characterize the pharmacokinetics of nicotine and cotinine, as well as 3′-OH cotinine and 3′-OH cotinine glucuronide, in adult and adolescent nonhuman primates. With some exceptions, the calibrated model predictions were in good agreement with experimental observations. Model performance (precision and bias)

Conclusions

In summary, a PBPK model was developed for the first time in adult and adolescent nonhuman primates to characterize the pharmacokinetics of nicotine and its metabolites, cotinine, 3’-OH cotinine, and 3’-OH cotinine glucuronide following intravenous and subcutaneous injections. The calibration of the current model suggests that adolescent nonhuman primates clear nicotine and cotinine more rapidly than adults. This may be explained by greater hepatic metabolic capacity in adolescent monkeys

Disclaimer

The findings and conclusions in this report are those of the authors and do not necessarily represent the official position of the Food and Drug Administration.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

The authors gratefully acknowledge the critical review of this manuscript by Drs. Luisa Camacho, Lydia Bilinsky, and Frederick Beland, FDA National Center for Toxicological Research. This study was funded by U.S. Food and Drug Administration, USA.

References (77)

  • J.G. Teeguarden et al.

    A multi-route model of nicotine-cotinine pharmacokinetics, pharmacodynamics and brain nicotinic acetylcholine receptor binding in humans

    Regul. Toxicol. Pharmacol.

    (2013)
  • X.X. Yang et al.

    Development of a physiologically based pharmacokinetic model for assessment of human exposure to bisphenol A

    Toxicol. Appl. Pharmacol.

    (2015)
  • N. Ageyama et al.

    Specific gravity of whole blood in cynomolgus monkeys (Macaca fascicularis), squirrel monkeys (Saimiri sciureus), and tamarins (Saguinus labiatus) and total blood volume in cynomolgus monkeys

    Contemp. Top. Lab. Anim. Sci.

    (2001)
  • T. Akabane et al.

    A comparison of pharmacokinetics between humans and monkeys

    Drug Metab. Dispos.

    (2010)
  • N. Al Koudsi et al.

    Hepatic CYP2A6 levels and nicotine metabolism: impact of genetic, physiological, environmental, and epigenetic factors

    Eur. J. Clin. Pharmacol.

    (2010)
  • M.C.R. Andrade et al.

    Biologic data of Macaca mulatta, Macaca fascicularis, and Saimiri sciureus used for research at the Fiocruz Primate Center

    Memorias Do Instituto Oswaldo Cruz

    (2004)
  • Z. Bao et al.

    Metabolism of nicotine and cotinine by human cytochrome P450 2A13

    Drug Metab. Dispos.

    (2005)
  • S. Bastianini et al.

    Long-term cardiovascular reprogramming by short-term perinatal exposure to nicotine’s main metabolite cotinine

    Acta Paediatr.

    (2018)
  • J. Beer et al.

    Maximum human lifespan may increase to 125 years

    Nature

    (2017)
  • J.D. Belluzzi et al.

    Age-dependent effects of nicotine on locomotor activity and conditioned place preference in rats

    Psychopharmacology

    (2004)
  • N.L. Benowitz

    Pharmacology of nicotine: addiction, smoking-induced disease, and therapeutics

    Annu. Rev. Pharmacol. Toxicol.

    (2009)
  • N.L. Benowitz et al.

    Interindividual variability in the metabolism and cardiovascular effects of nicotine in man

    J. Pharmacol. Exp. Ther.

    (1982)
  • N.L. Benowitz et al.

    Cotinine disposition and effects

    Clin. Pharmacol. Ther.

    (1983)
  • N.L. Benowitz et al.

    Female sex and oral contraceptive use accelerate nicotine metabolism

    Clin. Pharmacol. Ther.

    (2006)
  • N.L. Benowitz et al.

    CYP2A6 genotype and the metabolism and disposition kinetics of nicotine

    Clin. Pharmacol. Ther.

    (2006)
  • N.L. Benowitz et al.

    Nicotine chemistry, metabolism, kinetics and biomarkers

    Handb. Exp. Pharmacol.

    (2009)
  • D.R. Bolter et al.

    Morphometric analysis of growth and development in wild-collected vervet monkeys (Cercopithecus aethiops), with implications for growth patterns in Old World monkeys, apes and humans

    Zool. Soc. Lond.

    (2003)
  • R.P. Brown et al.

    Physiological parameter values for physiologically based pharmacokinetic models

    Toxicol. Ind. Health

    (1997)
  • J. Chen et al.

    Age of smoking initiation: implications for quitting

    Health Rep.

    (1998)
  • H.J. Clewell et al.

    Evaluation of the potential impact of age- and gender-specific pharmacokinetic differences on tissue dosimetry

    Toxicol. Sci.

    (2004)
  • M. Clozel et al.

    Effects of endothelin on regional blood flows in squirrel monkeys

    J. Pharmacol. Exp. Ther.

    (1989)
  • A.M. Collier et al.

    Cotinine elimination and its use as a biomarker in young children involuntarily exposed to environmental tobacco smoke

    Indoor Environ.

    (1994)
  • D.S. Counotte et al.

    Adolescent nicotine exposure transiently increases high-affinity nicotinic receptors and modulates inhibitory synaptic transmission in rat medial prefrontal cortex

    FASEB J.

    (2012)
  • E.L. Craig et al.

    Nicotine pharmacokinetics in rats is altered as a function of age, impacting the interpretation of animal model data

    Drug Metab. Dispos.

    (2014)
  • K.P. Davy et al.

    Total blood-volume in healthy-young and older men

    J. Appl. Physiol.

    (1994)
  • D. Dempsey et al.

    Nicotine metabolism and elimination kinetics in newborns

    Clin. Pharmacol. Ther.

    (2000)
  • D.A. Dempsey et al.

    CYP2A6 genotype but not age determines cotinine half-life in infants and children

    Clin. Pharmacol. Ther.

    (2013)
  • J.M. Drazen et al.

    Cardiac output at rest in the squirrel monkey: role of -adrenergic activity

    Am. J. Phys.

    (1972)
  • Cited by (2)

    • Deconvolution of Systemic Pharmacokinetics Predicts Inhaled Aerosol Dosimetry of Nicotine

      2023, European Journal of Pharmaceutical Sciences
      Citation Excerpt :

      However, nicotine is also known to bind to receptors, and earlier PBPK models developed for different routes of administration have incorporated receptor binding to predict tissue distribution (Plowchalk et al., 1992; Robinson et al., 2005; Rostami et al., 2022; Teeguarden et al., 2013; Yang et al., 2020). These PBPK models were able to capture the rapid clearance of high systemic nicotine concentrations in different species but were unable to describe the terminal plasma nicotine concentrations (Yang et al., 2020). Although the chemical properties of nicotine indicate lysosomal distribution as the primary mechanism influencing tissue distribution, the fractional contribution of each mechanism (i.e., lysosomal distribution and receptor binding) needs to be further evaluated.

    1

    Present address: Division of Nonclinical Science, Center for Tobacco Products, U.S. Food and Drug Administration, Silver Spring, MD, 20993, U.S.A.

    2

    Present address: Arkansas Center for Birth Defects Research, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72,202, U.S.A.

    3

    Present address: Arkansas Children's Research Institute, Little Rock AR 72202

    4

    Present address: Division of Behavioral Biology, School of Medicine, Johns Hopkins University, Baltimore, MD 21224, U.S.A.

    5

    Present address: Thermo Fisher Scientific, San Jose, CA, 95134, U.S.A.

    6

    Present address: Covance Inc., Salt Lake City, UT 84124, U.S.A.

    View full text